Depression as an Evolutionary Strategy for Defense Against Infection

Embed Size (px)

Citation preview

  • 8/20/2019 Depression as an Evolutionary Strategy for Defense Against Infection

    1/14

    Review

    Depression as an evolutionary strategy for defense against infection

    Sherry Anders a, Midori Tanaka b, Dennis K. Kinney c,d,⇑

    a Clinical Psychologist in Independent Practice, Boxborough, MA, USAb Obara Hospital, Sapporo, Hokkaido, Japanc McLean Hospital, Belmont, MA, USAd Department of Psychiatry, Harvard Medical School, Boston, MA, USA

    a r t i c l e i n f o

     Article history:

    Available online 20 December 2012

    Keywords:

    Depression

    Evolution

    Mood

    Inflammation

    Infection

    Infection-defense

    Immune

    Hypothesis

    Genetic

    Anti-depressants

    a b s t r a c t

    Recent discoveries relating depression to inflammation and immune function may help to solve an

    important evolutionary puzzle: If depression carries with it so many negative consequences, including

    notable costs to survival and reproduction, then why is it common and heritable? What countervailing

    force or compensatory advantage has allowed susceptibility genes for depression to persist in the popu-

    lation at such high rates? A priori, compensatory advantages in combating infection are a promising can-

    didate, given that infection has been the major cause of mortality throughout human history. Emerging

    evidence on deeply rooted bidirectional pathways of communication between the nervous and immune

    systems further supports this notion. Here we present an updated review of the ‘‘infection-defense

    hypothesis’’ of depression, which proposes that moods—with their ability to orchestrate a wide array

    of physical and behavioral responses—have played an adaptive role throughout human history by helping

    individuals fight existing infections, as well as helping both individuals and their kin avoid new ones. We

    discuss new evidence that supports several key predictions derived from the hypothesis, and compare it

    with other major evolutionary theories of depression. Specifically, we discuss how the infection-defense

    hypothesis helps to explain emerging data on psychoimmunological features of depression, as well as

    depression’s associations with a diverse array of conditions and illnesses—including nutritional deficien-

    cies, seasonal changes, hormonal fluctuations, and chronic diseases—that previous evolutionary theoriesof depression have not accounted for. Finally, we note the potential implications of the hypothesis for the

    treatment and prevention of depression.

     2012 Elsevier Inc. All rights reserved.

    1. Introduction: rationale for an ‘‘infection-defense’’ hypothesis

    of depression

    Infection has been the leading cause of mortality throughout

    human history (Cairns, 1997; Finch, 2010). It has been estimated

    that prior to the industrial period, the average life expectancy

    was 25, and it was not uncommon for half of the siblings in a fam-

    ily to die before reaching adulthood (Cairns, 1997; Casanova and

    Abel, 2005). Particularly virulent pathogens could wipe out an en-tire family or village, such as the English ‘‘sweating sickness’’

    known to have wiped out one-half to two-thirds of the population

    in many English towns during the late 1400s and early 1500s

    (Thwaites et al., 1997). With such stark capabilities, infection has

    been a critical and potent driving force in natural selection

    (Dobson and Carper, 1996). Specific alleles have evolved in re-

    sponse to common pathogens in an environment; however, patho-

    gens are ubiquitous and wide-ranging, with new forms continually

    evolving, leaving individuals intrinsically vulnerable (Casanova

    and Abel, 2005; Dobson and Carper, 1996). The ideal system of de-

    fense against this inherent vulnerability to infection requires a

    generalized response that is proactive in reducing infection risk

    during times of increased vulnerability, as well as both flexible

    and adaptive enough to provide resistance to a wide range of 

    pathogens.

    Here, and in several recent papers (Kinney and Tanaka, 2009;

    Tanaka and Kinney, 2011a,b; Tanaka et al., 2012), we propose thatmoods—with their ability to orchestrate a wide array of physical

    and behavioral responses—have evolved as part of a complex sys-

    tem of immune defense that helps counteract our inherent vulner-

    ability to the diversity of environmental pathogens. The ‘‘infection-

    defense hypothesis’’ offers a novel evolutionary framework for

    understanding how many of the social and behavioral features of 

    depression may help individuals fight existing infections, as well

    as help both individuals and their family members avoid new ones.

    In contrast to many previous evolutionary theories of depression, it

    takes into account and helps to integrate a large and growing body

    of evidence linking depression to inflammation and immune func-

    tion, and helps to explain depression’s association with a vast array

    0889-1591/$ - see front matter  2012 Elsevier Inc. All rights reserved.http://dx.doi.org/10.1016/j.bbi.2012.12.002

    ⇑ Corresponding author. Address: 18 Locust Avenue, Lexington, MA 02421, USA

    Tel.: +1 781 862 1644, mobile: +1 617 271 5156; fax: +1 781 862 6559.

    E-mail address:  [email protected] (D.K. Kinney).

    Brain, Behavior, and Immunity 31 (2013) 9–22

    Contents lists available at SciVerse ScienceDirect

    Brain, Behavior, and Immunity

    j o u r n a l h o m e p a g e :   w w w . e l s e v i e r . c o m / l o c a t e / y b r b i

    http://dx.doi.org/10.1016/j.bbi.2012.12.002mailto:[email protected]://dx.doi.org/10.1016/j.bbi.2012.12.002http://www.sciencedirect.com/science/journal/08891591http://www.elsevier.com/locate/ybrbihttp://www.elsevier.com/locate/ybrbihttp://www.sciencedirect.com/science/journal/08891591http://dx.doi.org/10.1016/j.bbi.2012.12.002mailto:[email protected]://dx.doi.org/10.1016/j.bbi.2012.12.002

  • 8/20/2019 Depression as an Evolutionary Strategy for Defense Against Infection

    2/14

    of conditions and illnesses such as nutritional deficiencies, sea-

    sonal changes, hormonal fluctuations, and chronic diseases. The

    infection-defense hypothesis may also help to resolve a baffling

    evolutionary puzzle—why major psychological depression is so

    prevalent and heritable, despite its high costs for survival and

    reproduction, including suicide (Tanaka and Kinney, 2011a).

    We begin the paper with an overview of the epidemiology and

    neurobiology of depression, including recent updates linking

    depression to immune factors. This is followed by a discussion of 

    the infection-defense hypothesis (Kinney and Tanaka, 2009), in

    which we propose that depressive features provide advantages in

    combating infectious diseases—advantages that offset many

    known disadvantages of depression. We also briefly review other

    hypotheses and evidence that have related changes in immune

    function to depression. We then describe several testable predic-

    tions of the ‘‘infection-defense hypothesis’’ and discuss empirical

    evidence that bears on each prediction. We conclude by comparing

    the hypothesis with other evolutionary theories of depression and

    by discussing some potential implications of the hypothesis for

    better treatment and prevention of depression.

    2. Depression: an evolutionary puzzle

    While depression can occur at any point during the lifespan, its

    most frequent onset occurs during the peak years of work and

    reproduction—and both depressed individuals and their family

    members often endure serious physical, social, and economic bur-

    dens as a result (Broadhead et al., 1990; Eaton et al., 1997; Kler-

    man, 1989). Depression negatively impacts productivity, as well

    as psychosocial functioning, and is associated with increased rates

    of unemployment and divorce (Weissman et al., 1996). Depression

    is associated with lower rates of fertility (Tondo et al., 2011; Wil-

    liams et al., 2007; Yates et al., 2010), and children of depressed

    mothers show poorer average outcomes on a wide range of devel-

    opmental indices (Cummings and Davies, 1994), with adverse con-

    sequences noted even in cases where there has only been prenatalexposure to maternal depression (Davis et al., 2007). Increased

    mortality rates are also associated with depression, as depression

    is a risk-factor for many disease-related causes of death as well

    as for suicide (Mykletun et al., 2007). From a global perspective,

    the World Health Organization (WHO) has made the projection

    that, by the year 2020, depression will be the 2nd leading cause

    of disease burden worldwide (Murray and Lopez, 1996).

    Depression thus poses a baffling evolutionary puzzle; despite

    the serious consequences of depression for individuals and their

    family members, including decreased fertility and increased mor-

    tality rates, depression remains both common and heritable. The

    estimated lifetime risk of a major depressive episode has risen to

    23% in the United States (Kessler et al., 2005), and there is evidence

    to suggest that the incidence of major depressive disorder mayactually be increasing (e.g.,   Compton et al., 2006). Moreover, the

    heritability of depression is well-established, with estimates based

    on twin, adoption, and genetic molecular studies consistently fall-

    ing at about 40% (Kendler et al., 1995; McGuffin et al., 1996; Shyn

    and Hamilton, 2010; Sullivan et al., 2000; Wender et al., 1986). Ge-

    netic association and linkage studies have begun to discover spe-

    cific alleles that increase risk for depression (see review by

    Goldberg, 2006), such the CYP2C9⁄3 allelle (LLerená et al., 2003)

    and the 5-HTTLPR short allele of the serotonin transporter gene

    (Eley et al., 2004; Kendler et al., 2005).

    For more than half a century, efforts to understand the neuro-

    biological underpinnings of depression have been dominated by

    the view that depression is caused by a deficiency in synaptic con-

    centrations of monoaminergic neurotransmitters including seroto-nin and norepinephrine (Hirschfeld, 2000; Schildkraut and Kety,

    1967). This idea, known as the monoamine hypothesis, has stimu-

    lated a wealth of research and has been the major driving force be-

    hind antidepressant drug development. Over time, however, the

    initial promise of the monoamine hypothesis has been tempered

    by the fact that attempts to find direct links between monoaminer-

    gic transmission and mood have yielded equivocal results (Delgad-

    o, 2000; Heninger et al., 1996). In addition, the efficacy of 

    antidepressant drugs based on the fundamental premise of the

    monoamine hypothesis has been limited, with estimates that be-

    tween 30% and 50% of individuals treated with antidepressant

    medication do not show adequate response (Schatzberg, 2000).

    The insufficiency of the monoamine hypothesis to explain critical

    aspects of mood regulation and the desire for more favorable treat-

    ment outcomes have resulted in an expanded search for the neuro-

    biological underpinnings of depression.

    Research on the neuroimmune system and its role in the etiol-

    ogy of depression has emerged as an especially promising area for

    study. In particular, the role of immune-activated inflammatory

    cytokines has been identified as a key area of focus in understand-

    ing the neurobiological pathways that trigger depressive states, by

    way of direct and indirect effects on hypothalamic–pituitary–adre-

    nal (HPA) axis, and by altering monoamine neurotransmitters in

    multiple regions of the brain (Dantzer et al., 2008; Loftis et al.,

    2010; Raison et al., 2006). In addition, a recent review of risk alleles

    for depression has revealed that in a striking majority of cases the

    depression alleles were associated with known effects on immune

    function (Raison and Miller, 2012). These new lines of research—

    which show wide ranging links between depressive symptomatol-

    ogy and immune function—not only have the potential to lead to

    novel treatment strategies for the prevention and treatment of 

    depression, but may also provide important clues as to the reasons

    for depression’s prevalence and persistence throughout human

    history.

    3. Immune alterations, mood, and the macrophage and

    cytokine theories of depression

    Numerous associations between depression and immune func-

    tion have been observed in recent years. Early studies investigating

    immune alterations during depression focused almost exclusively

    on markers of suppressed cellular immunity—such as decreased

    lymphocyte proliferation and natural killer (NK) cell activity (Rei-

    che et al., 2004; Schleifer et al., 1989; Zorilla et al., 2001). More re-

    cently, however, there has been a shift toward understanding the

    role of inflammation in depression, with a particular focus on the

    role of proinflammatory cytokines (Zorilla et al., 2001; Irwin and

    Miller, 2007). Increased levels of proinflammatory cytokines—such

    as interleukin-1b   (IL-1b), interleukin-6 (IL-6), tumor necrosis fac-

    tor-a (TNF-a), and interferon-c (IFN-c)—have been repeatedly ob-served in depressed individuals, prompting formulation of the

    ‘macrophage theory of depression’ (Smith, 1991), and its succes-

    sive formulation as the ‘cytokine hypothesis of depression’ (Maes

    et al., 1995; Maes, 1999; Raison et al., 2006; Schiepers et al.,

    2005). According to the cytokine hypothesis, proinflammatory

    cytokines produced by macrophages during the acute phase of an

    immune response act as neuromodulators that mediate the behav-

    ioral and neurobiological features of depression.

     3.1. Cytokine-induced changes in somatic experience, cognition, and

    behavior 

    When infection or injury occurs, proinflammatory cytokines are

    responsible for orchestrating the early immune response, including

    sickness behavior. Sickness behavior—characterized by somatic,cognitive, and behavioral changes, such as fever, weakness, mal-

    10   S. Anders et al. / Brain, Behavior, and Immunity 31 (2013) 9–22

  • 8/20/2019 Depression as an Evolutionary Strategy for Defense Against Infection

    3/14

    aise, listlessness, hyperalgesia, and impaired concentration (Hart,

    1988)—represents an organized strategy for fighting infection by

    conserving metabolic resources and helping an individual avoid

    further stressors (Hart, 1988; Kluger, 1991; Segerstrom, 2010; Yir-

    miya et al., 2000). Dantzer (2001, 2009) has extended this notion to

    suggest that sickness behavior is an expression of a biologically-

    mediated motivational state triggered by the innate immune sys-

    tem that resets an organism’s priorities to adaptively cope with

    the threat of bodily insult. The costs of shifting resources and pri-

    orities during this state are purportedly offset by the critical advan-

    tages offered for fighting infection.

    A portion of individuals who experience sickness behavior go on

    to develop major depressive disorder. There is evidence that sec-

    ondary development of depression following sickness behavior oc-

    curs in individuals who have an exaggerated vulnerability to

    infection (Dantzer, 2009; Wichers et al., 2006). For example, the

    therapeutic administration of cytokines such as interferon-a(IFN-a) and interleukin-2 (IL-2)—used to treat patients with cancerand a number of infectious diseases—has been found to induce a

    two-phase response. First, a few days following cytokine adminis-

    tration, many patients experience sickness behavior, marked by

    neurovegetative and somatic symptoms (fatigue, aches, loss of 

    appetite, sleep disturbance). Second, up to 50% of those treated

    go on to develop symptoms of major depression, including de-

    pressed mood, feelings of worthlessness, guilt, and even suicidal

    ideation (Capuron et al., 2002; Capuron and Miller, 2004). Individ-

    uals who are most likely to develop depression during the course of 

    cytokine therapy have been found to show increased baseline lev-

    els of proinflammatory cytokines prior to treatment (Wichers et al.,

    2006), and more severe levels of sickness-type behavior following

    initial administration of treatment (Robaeys et al., 2007; Wichers

    et al., 2005). Notably, cytokine-induced symptoms of depression

    are attenuated by pretreatment with antidepressant therapy

    (Capuron et al., 2002).

     3.2. Sickness behavior and depression compared

    There are many parallels between sickness behavior and

    depression, although the two conditions are not equivalent (see Ta-

    ble 1). For example, similar to sickness behavior, symptoms of 

    depression such as anhedonia, fatigue, hypersomnia, and psycho-

    motor retardation (i.e., slowed speech, thinking, and body move-

    ments) all tend to reduce activity and encourage rest, thereby

    conserving energy. Some studies have found that depression is

    associated with mild elevations in body temperature (e.g.,   Avery

    et al., 1999; Rausch et al., 2003), and there is suggestive evidence

    that milder temperature elevations, particularly when combined

    with reduced bodily iron stores, may be inhibitive to pathogen

    growth (Ismael and Bedell, 1986; Kluger and Rothenburg, 1979).

    Nevertheless, a major difference between sickness behavior and

    depression is that the costly antibiotic strategy of fever is notably

    subverted during depression (Maier and Watkins, 1998). Depres-

    sion also differs from sickness behavior in that its symptoms are

    more variable and include behavioral and cognitive features that

    discourage social contact and activity. Several authors have sug-

    gested that depressive symptoms, like sickness behavior, may not

     just be a spurious byproduct of cytokine-induced inflammation,

    but may constitute an adaptive longer-term strategy for fighting

    infection (Raison et al., 2006; Kinney and Tanaka, 2009).

     3.3. Neuromodulatory mechanisms of cytokines

    Multiple pathways have been identified by which cytokines ex-

    ert neuromodulatory effects, and these have been covered exten-

    sively in a number of recent reviews (e.g.,   Capuron and Miller,

    2011; Dantzer et al., 2008; Maes et al., 2009; Miller et al., 2009;

    Loftis et al., 2010). Proinflammatory cytokines have been noted,for example, to exert both direct and indirect effects on mono-

    amine neurotransmitter availability and metabolism (Dunn and

    Wang, 1995), as well as to increase activation of the hypotha-

    lamic–pituitary–adrenal (HPA) axis (Holsboer, 2000; Pace et al.,

    2007). A number of recent studies have also demonstrated links

    between inflammatory activation and brain changes that underlie

    depression (e.g., Brydon et al., 2008; Eisenberger et al., 2010; Har-

    rison et al., 2009).

    Cytokines administered to laboratory animals and humans have

    been shown to alter the metabolism of the neurotransmitters sero-

    tonin, norepinephrine, and dopamine in areas of the brain that are

    associated with mood regulation (Rivier et al., 1989; Shuto et al.,

    1997; Capuron et al., 2003). A number of studies suggest that cyto-

    kines may decrease the amount of serotonin available for neuro-transmission by up-regulating the expression and activity of the

    serotonin transporter (SERT) (Katafuchi et al., 2006; Zhu et al.,

    2006). One of the most studied pathways by which cytokines

    may influence neurotransmitter metabolism, however, involves

    degradation of tryptophan by activation of the enzyme, indole-

    amine 2,3-dioxygenase (IDO) (Capuron et al., 2003; Dantzer

    et al., 2008). IDO is activated by proinflammatory cytokines

    released during the acute-phase immune response, including

    IFN-a, IL-6, and TNF. Serotonin levels are reduced due to the

     Table 1

    A comparison of sickness behavior and depression based on the ‘‘infection-defense hypothesis’’.

    Sickness behavior Depression

    Eliciting Conditions   Infection, trauma, injury Immune vulnerability, immune compromise

    Mode of Action   Reactive Proactive and/or complementary; may act preemptively to avoid infection

    during times of increased immune vulnerability, or help combat existing infections

    Symptoms   Circumscribed, including fever, malaise, muscle

    and joint aches, impaired concentration, fatigue,

    loss of appetite

    Variable, includingsadness, loss of interest and pleasure, lowmotivation, withdrawal,

    impaired concentration, fatigue, decreased sex drive, decreased speech, psychomotor

    slowing, appetite disturbance, sleep disturbance, low self- esteem, hopelessness,

    guilt, suicidal ideation, and somatic changes such as increased pain sensitivity, aches,

    and mild hyperthermia.

    Depressive symptoms are diverse and vary across patients and subtypes (e.g.,

    melancholia may involve hypervigilance with agitation and insomnia that is less

    common in atypical, postpartum, or seasonal subtypes); however, nearly all

    symptoms appear to help fight or avoid infection in some way.

    Duration   Acute; sustained activation is biologically

    costly and harmful to host

    Sustainable for indefinite periods of time

    Inflammatory Response   Sickness behavior always involves inflammation Depression commonly, but not necessarily, involves inflammation

     Adaptive Function   Acute mobilization of resources to combat

    infection and promote healing

    Help combat existing infections, help avoid new immune-compromising stressors,

    and help individuals and their kin avoid new infections

    S. Anders et al. / Brain, Behavior, and Immunity 31 (2013) 9–22   11

  • 8/20/2019 Depression as an Evolutionary Strategy for Defense Against Infection

    4/14

    breakdown of tryptophan by IDO, resulting in the production of 

    several neurotoxic compounds that may further mediate a depres-

    sive response (Dantzer et al., 2008). IDO activity diverts tryptophan

    metabolism from the production of serotonin to the synthesis of its

    primary metabolite kynurenine, which is further metabolized into

    several neuroactive compounds that include 3-hydroxykynurenine

    (3-HK) and quinolinic acid (QUIN) or kynurenic acid (KA). These

    compounds, in turn, generate free radicals that cause neuronal

    damage due to oxidative stress (Wichers and Maes, 2004). The po-

    tential importance of kynurenine metabolites in inflammatory-

    mediated depression is underscored by the results of a recent

    study in which Raison et al. (2010) found that individuals receiving

    IFN-a for treatment of Hepatitis C were found to have higher cere-brospinal fluid (CSF) levels of kynurenine and its metabolites QUIN

    and KA, and that these increases were correlated with increased

    inflammatory biomarkers as well as depression.

    Depression also has well-established links with hyperactivity of 

    the HPA-axis (Pariante and Lightman, 2008). Cytokines may con-

    tribute to depression either directly, via activation of the HPA-axis,

    or indirectly, through cytokine-induced glucocorticoid-receptor

    resistance (Holsboer, 2000; Pace et al., 2007). Cytokines activate

    the HPA-axis by inducing corticotropin-releasing hormone (CRH)

    and vasopressin (AVP)—key regulators of the HPA-axis that are

    found at increased levels in depressed individuals (see  Owens

    and Nemeroff, 1991; Holsboer, 2000; Scott and Dinan, 2002). Cyto-

    kines may contribute to HPA-axis hyperactivity indirectly, as well,

    through their effects on the glucocorticoid receptor. Cytokines in-

    crease glucocorticoid receptor resistance by way of several signal-

    ing pathways, including activation of the p38 MAPK, and by

    stimulating changes in the expression of glucocorticoid receptor

    isoforms (Pace et al., 2007). These changes, in turn, create a dysreg-

    ulation of the CRH feedback system and result in a feed-forward

    cascade that decreases the inhibitory effect of glucocorticoids on

    CRH and stimulates increased cytokine production.

    Further evidence on the neuromeodulatory effects of cytokines

    comes from studies using functional magnetic resonance imaging

    (fMRI) techniques. A number of studies have shown that inflamma-tory activation can stimulate changes in key areas of the brain that

    underlie depression. For example, following experimental induc-

    tion of an immune-inflammatory response,   Eisenberger et al.

    (2010) found that research participants showed increased symp-

    toms of depression, and reduced activity in the ventral striatum

    (VS)—the neural correlate of anhedonia—in response to reward

    cues. In another study investigating the neural correlates of psy-

    chomotor slowing, neural activity in the substantia nigra was

    found to correspond to increased levels of IL-6 and decreased reac-

    tion times following immune stimulation (Brydon et al., 2008). The

    same research group has also reported findings that depressed

    mood and increased circulating levels of IL-6 following immune

    stimulation correspond to changes in areas of the brain highly in-

    volved in emotional processing. Specifically, they found increasedsubgenual anterior cingulate cortex (sACC) activity and reduced

    connectivity of sACC to mesolimbic regions of the brain including

    the amygdala, medial prefrontal cortex, nucleus accumbens, and

    superior temporal sulcus (Harrison et al., 2009).

    4. The ‘‘infection-defense hypothesis’’ of depression

    Advances in understanding the ways in which immune func-

    tion, inflammation, and depression are related potentially provide

    clues as to why genes that increase vulnerability to depression

    have persisted at relatively high levels in the gene pool, despite

    depression’s significant costs to reproduction and survival. The

    infection-defense hypothesis of depression (Kinney and Tanaka,2009) offers an integrative view of these findings, based on the idea

    that depression confers a critical compensatory advantage in im-

    mune protection that has offset notable disadvantages of depres-

    sion for evolutionary fitness. More specifically, the infection-

    defense hypothesis proposes that immune vulnerability to infec-

    tion elicits depressed mood, which in turn stimulates behaviors

    that help protect vulnerable individuals and their kin against infec-

    tious diseases (see Table 2).

    The immune system is an amazingly sophisticated system

    involving multiple, layered, and complementary strategies and

    mechanisms for helping individuals combat infections, second only

    in complexity to the nervous system. That natural selection has

    sculpted this incredibly complex system is testament to what a

    powerful role infection has played in natural selection. What we

    are hypothesizing here is that natural selection may have also

    sculpted coordinated pathways between the nervous and immune

    systems so as to evolve behavioral response mechanisms that help

    prevent and combat infections. Others, including Hart, have noted

    the potential for such mechanisms: ‘‘It is quite logical to expect

    animals and people to also have evolved nonimmunologic dis-

    ease-fighting strategies including behavioral patterns, that might

    serve as a first line of defense before the nonspecific and specific

    immunologic systems are activated and that would complement

    or potentiate immunological processes’’ (Hart, 1988 (p. 123)). As

    Hart notes, behavioral responses, in principle, offer a valuable po-

    tential advantage for combating infection—which is to avoid

    becoming infected in the first place. There are numerous examples

    of preventive immune strategies including, for example, human

    aversion to noxious odors, which helps to prevent contact withhigh-pathogen sources such as human waste and decaying flesh.

    Although at first glance it may seem counter-intuitive, from an

    evolutionary perspective, it is precisely the high costs associated

    with depression, combined with the fact that it is common and

    heritable, that argues for some adaptive advantage to depression.

    That is, given such negative consequences, there must be some

    critical countervailing force or compensatory advantage that has

    allowed susceptibility genes for depression to remain in the popu-

    lation at high rates. Compensatory advantages in combating infec-

    tion are a promising candidate, given that infection has had strong

    associations with morbidity and mortality throughout evolution-

    ary history. Increasing evidence that complex, deeply rooted mech-

    anisms have evolved, by which the nervous and immune systems

    can communicate and influence one another, also points in thisdirection.

    Examples whereby costly adaptations have evolved because

    they offer compensatory advantages in defending against infection

    are numerous and widespread. Classic examples in humans in-

     Table 2

    Key postulates of the infection-defense hypothesis of depression.

    1. Immune vulnerability to infectionelicits depressed mood, which in turnstimulates behaviors that help protect vulnerable individuals and their kin against infectious

    diseases.

    2. Depressive signs and symptoms offer individuals advantages for fighting existing infections by helping to conserve energy and avoid environmental stressors and

    insults that could provide further immune-compromising challenge.

    3. By inhibiting social contact, depression also prevents individuals and their biological relatives from contracting new infections.

    4. Moods orchestrate—in a timely, titrated, and strategic manner—an array of behavioral and immunological responses to infections and immune vulnerability, which

    can in turn be affected by a wide range of factors, including, e.g., genetic and seasonal variables, physical illness or injury, nutritional status, hormonal fluctuations,

    exposure to environmental toxins, sleep disturbance, and stress.

    12   S. Anders et al. / Brain, Behavior, and Immunity 31 (2013) 9–22

  • 8/20/2019 Depression as an Evolutionary Strategy for Defense Against Infection

    5/14

    clude sickle cell anemia and other hereditary hemoglobinopathies,

    such as thallasemia. Sickle cell anemia, for example, is a hereditary

    disease that is typically fatal, yet it is very prevalent in tropical re-

    gions. People who inherit only one copy of the sickle-cell gene have

    increased resistance to malaria, allowing the gene to persist de-

    spite its severe costs (Kwiatkowski, 2005). An example from non-

    human species involves the evolution of eye-catching plumage

    and courtship dances that peacocks and the males of many other

    avian species use to woo females. At first glance—from an evolu-

    tionary perspective—this is a rather puzzling phenomenon; after

    all, it takes significant energy and metabolic resources for the birds

    to produce those ostentatious feathers and dances, and increases

    visibility to predators. So why do it? Field studies indicate that it

    is adaptive because showy feathers and skillful dance signal to

    the females that the male carries fewer parasites. When the female

    chooses the visually more attractive males, she reduces her own

    exposure (and that of her offspring) to infection, and increases

    the likelihood that the male (and her offspring) will have better ge-

    netic resistance to infections in that ecological niche (see  Kempe-

    naers, 2007).

    Behavioral strategies that defend kin against risk of infection

    can also be noted in species such as honey bees and African naked

    mole rats (see Preti, 2007). These species are like humans in that

    they are social and live together in large, complex communities,

    and have been known to display altruistic self-sacrifice, often leav-

    ing the hive or burrow to die when they are ill—reducing the risk

    that they will expose others within their community to the

    infection.

    From an evolutionary perspective, moods potentially provide an

    attractive system for behavioral defense against infection, as they

    have the ability to (1) orchestrate a wide range of behaviors; (2)

    create responses that are generalizable to a variety of environmen-

    tal challenges—a variety that reflects the inherent mutability and

    variability of environmental pathogens; (3) respond to immune

    vulnerability in a way that is both timely and titrated; and (4)

    act preemptively, based on both endogenous and environmental

    signals of increased infection risk. Depression’s overlapping fea-tures with sickness behavior appear to help conserve energy and

    avoid further immune challenges, while depressive states are more

    sustainable than sickness behavior over longer periods of time by

    reducing the high cost of fever. This may allow depression to serve

    as a complementary line of defense when the early immune re-

    sponse to infection cannot be sustained during extended periods

    of infection or immune vulnerability.

    What we propose is even broader, however. A key aspect that

    distinguishes the infection-defense hypothesis from many other

    evolutionary theories of depression is that it proposes that depres-

    sive signs and symptoms not only offer individuals advantages for

    fighting existing infections, but also for preventing individuals and

    their biological relatives from contracting new infections. Social

    withdrawal, low energy, irritability, and blunted affect, for exam-ple, may greatly reduce the spread of infections by discouraging

    mobility and/or social contact with others.

    5. Some testable predictions of the infection-defense hypothesis

    A number of testable predictions follow from the infection-de-

    fense hypothesis. These predictions include the following:

    1. Most signs and symptoms of depression will aid the immune

    system’s ability to fight infections, by performing one or more

    of the following functions:

    (a) conserving metabolic resources for use by the immune sys-

    tem in fighting infection;

    (b) directly enhancing immune function through antimicrobialaction or by stimulating an increase in NK cell activity;

    (c) reducing the risk of further environmental stresses that

    impair immune function; and/or

    (d) helping individuals and their kin to avoid transmitting or

    contracting new infections.

    2. Many types of infectious diseases will be associated with

    depressive symptoms.

    3. Depressed individuals will tend to have elevated rates of infec-

    tion and/or immune alteration.

    4. Medical, environmental, and physiological conditions that

    increase immune vulnerability, or that increase exposure to

    infection, will also be associated with increased rates of 

    depression.

    5. There are bidirectional processes that communicate between

    the nervous and immune systems and provide mechanisms

    for infections, immune processes, and mood to influence one

    another.

    6. Moods provide an implicit mechanism for cost-benefit analysis

    of an individual’s optimal responses to environmental chal-

    lenges and the organisms’ immune status, helping to regulate

    the timing and intensity of infection-defense responses.

    Considerable evidence exists to support each of these predic-

    tions. Evidence relevant to the respective predictions is discussed

    in the following six sections.

    6. Evidence for prediction # 1: most signs and symptoms of 

    depression appear to aid the immune system’s ability to fight

    infections

    As the infection-defense hypothesis predicts, there is evidence

    that most features of depression appear to aid defense against

    infections. Depressive signs and symptoms do this in four ways:

    (a) conserving energy; (b) directly enhancing immune function

    through antimicrobial action or by stimulating an increase in NK

    cell activity; (c) reducing the risk of further immune-compromis-

    ing challenges; and/or (d) by helping individuals and their kinavoid contracting new infections (see Fig. 1).

    Akin to the symptoms of sickness behavior present during an

    acute immune response to infection, depressive symptoms—such

    as increased bodily aches and fatigue, hypersomnia, and psycho-

    motor retardation—also reduce physical activity and encourage

    rest, thereby conserving metabolic resources for use by the im-

    mune system. In addition to these somatic and behavioral changes

    that occur during depression, anhedonia and cognitive features of 

    depression—such as decreased self-esteem and feelings of hope-

    lessness (American Psychiatric Association, 2000)—also discourage

    exploration and risk-taking, thereby reducing exposure to new

    pathogens or to accidents, conflicts, or injuries that can exacerbate

    existing immune system compromise. A number of studies have

    found, for example, that depressed individuals exhibit state-dependent increases in harm-avoidance behavior (e.g.,  de Winter

    et al., 2007; Nery et al., 2009). Moreover, the degree of harm-avoid-

    ance behavior endorsed by depressed individuals increases along

    with increases in depression severity and the number of depressive

    episodes (Nery et al., 2009).

    Moods help to regulate social behavior. Social withdrawal and

    decreased sexual drive, as well as cues that discourage social con-

    tact from others—such as decreased speech, irritability, and

    changes in body language and facial expression (blunted affect)—

    can reduce the risk for contracting or transmitting infections,

    including sexually transmitted ones. Thus, the changes in social

    interest and demeanor associated with depression not only reduce

    an individual’s risk for contracting new infections by discouraging

    close contact with others, but also reduce the risk of transmittinginfection to relatives. The importance of reducing the spread of 

    S. Anders et al. / Brain, Behavior, and Immunity 31 (2013) 9–22   13

  • 8/20/2019 Depression as an Evolutionary Strategy for Defense Against Infection

    6/14

    infection to others is highlighted by many examples throughout

    human history whereby infectious diseases were known to wipe

    out entire families or villages (e.g., Thwaites et al., 1997). The nas-

    cent immune system of infants and young children render them

    especially vulnerable to infections. Consistent with the notion of 

    a heightened need to protect the young, depression among human

    mothers, as well as sickness behavior among murine animal mod-

    els, is associated with disengagement—and sometimes even rejec-tion—of offspring (Burke, 2003; Dantzer, 2009).

    Reduced mobility and reduced sociability associated with

    depression, as noted by Raison and Miller (2012), may also have

    the effect of minimizing exposure to out-group members. Contact

    with individuals from different groups or environments who are

    immunologically dissimilar increases the risk of exposure to patho-

    gens to which an individual has not developed specific immunity

    in their home environment.

    Appetite changes are extremely common in depression. De-

    pressed individuals can vary in whether they tend to increase or

    decrease overall intake of food (American Psychiatric Association,

    2000). However, the specific forms that these appetite changes

    take in depression may constitute alternative strategies that help

    defend against infection. Proinflammatory cytokines, for example,have a number of known anorectic effects, including increased

    stimulation of the peptide leptin, which plays a key regulatory role

    in balancing food intake and energy expenditure (Andréasson et al.,

    2007; Wong and Pinkney, 2004). Reduced appetite can reduce the

    risk of food–borne parasites, a major source of infection through-

    out history and the present (Mead et al., 1999). By contrast, some

    forms of depression, including seasonal affective disorder (SAD),

    are associated with increased carbohydrate cravings. Carbohy-

    drates and carbohydrate-based foods, such as sugar and bakedbread, offer high metabolic and caloric resources, while at the same

    time being less likely to cause illness due to spoiling than many

    other foods, such as meat, fish, or fruits and vegetables. In addition,

    increased intake of carbohydrates influences immune function by

    triggering changes in proinflammatory cytokines and an increase

    in NK cell activity (see Braun and Von Duvillard, 2004). It has also

    been noted that high intake of carbohydrates can promote defense

    against infection by decreasing the relative intake of lipids, as lipid

    consumption has been associated with worse outcomes for infec-

    tious illness (Heyland et al., 1998).

    Some individuals with depression develop paradoxical symp-

    toms that include notable anxiety, agitation, and insomnia, consti-

    tuting a state of hypervigilance. In spite of high metabolic costs,

    heightened vigilance is likely to have been an extremely adaptiveresponse to stress throughout human evolution, where the threat

    Fig. 1.   A neuro-immune feedback system regulates the timing and intensity of behaviors that aid defense against infection. Immune vulnerability is influenced by acute risk

    of infection, as well as by a number of other known immune-compromising factors that include stress, sleep deprivation, chronic infection and disease, winter season,

    hormonal fluctuations, exposure to toxins, and nutritional deficiencies. According to the infection-defense hypothesis, depression and related behaviors that aid against

    infection are activated by immune status, which in turn reduce immune vulnerability and risk of infection by (a) conserving or enhancing metabolic resources for fightinginfection, (b) directly aiding immune defenses through antimicrobial action and/or by increasing NK cell activity, (c) reducing risk of further immune challenge, such as

    avoiding danger or conflict to reduce the risk of physical injury, or restricting patterns of eating to avoid food-borne illness, and/or (d) by reducing the spread of infection

    through decreased social contact.  1 Depressive symptoms vary by subtype, accounting for seemingly paradoxical symptoms involving energy and appetite. For example,

    seasonal affective disorder (SAD) and other atypical forms of depressionoften include symptoms of psychomotor slowing, hypersomnia, and increased appetite/carbohydrate

    cravings, while other forms of depression – such as melancholia – include symptoms of restless agitation, anorexia, and insomnia.

    14   S. Anders et al. / Brain, Behavior, and Immunity 31 (2013) 9–22

  • 8/20/2019 Depression as an Evolutionary Strategy for Defense Against Infection

    7/14

    of predators and other physical dangers to self and family was high

    (Marks and Nesse, 1994). Hypervigilance may contribute to harm-

    avoidance behaviors that, as noted earlier, reduce risk of exposure

    to new infections or immune-compromising injuries and stressors.

    Furthermore, although speculative, restless pacing or hand-wring-

    ing, which can occur during agitated forms of depression, may offer

    some benefits to immune function just as other forms of moderate

    physical activity do. For example, moderate physical activity can

    increase NK cell activity (Nieman et al., 2005) and elevate core

    body temperature, which may have additional immune effects

    (Mestre-Alfaro et al., 2012). An agitated state tends to occur in

    the context of patients’ increased anxiety about their personal

    safety and that of their family, thereby discouraging them from

    adventurous activity outside the home, so that the increased exer-

    cise occurs in a way that reduces the risk of injury and contact with

    new infections that would otherwise tend to occur as a result of in-

    creased exercise.

    7. Evidence for prediction # 2: many types of infection are

    associated with depression

    Increased rates of depressive symptoms have been observed

    following an individual’s contracting a number of acute or suba-

    cute infections (Fazekas et al., 2006; Murray et al., 2007), as well

    as following chronic infections (Cohen et al., 2002; Lipkin and Hor-

    nig, 2004; O’Connor et al., 2009; Yates and Gleason, 1998), and

    immunization with live virus vaccines (Afsar et al., 2009; Glaser

    et al., 2003; Yirmiya et al., 2000). For example, increased depres-

    sive symptoms have been found in patients with herpes simplex

    encephalitis (Fazekas et al., 2006) and West Nile virus (WNV)

    (Murray et al., 2007). Murray et al. (2007) found that over 31% of 

    WNV patients in their study experienced the onset of a depressive

    episode within one year of their becoming infected. Those who had

    been diagnosed with the more severe form of WNV, neuroinvasive

    disease, showed an even greater risk for developing depression.

    Evidence for an increased risk of depression among those whosuffer from chronic infections comes from a variety of studies

    involving both humans and laboratory animals. Depression-like

    behavior has been observed, for example, in mice infected with Ba-

    cille Calmette Guerin (BCG), a bacterium related to the one that

    causes tuberculosis (O’Connor et al., 2009). Humans suffering from

    chronic infections, such as human immunodeficiency virus (HIV),

    hepatitis C (HCV), and genital herpes (HSV-2), have also been found

    to have an increased risk for depression (Yates and Gleason, 1998;

    Lipkin and Hornig, 2004). Among HIV patients, high viral loads are

    associated with higher depression scores on the Hamilton Rating

    Scale for Depression (Cohen et al., 2002).

    Vaccines containing live viruses can also cause depressive

    symptoms. For example, in a prospective double-blind study (Yir-

    miya et al., 2000), vaccination with live attenuated rubella virus re-sulted in depressive symptoms that lasted up to 10 weeks. In

    another study, Afsar et al. (2009)  found increases in symptoms of 

    depression related to antibody response following Hepatitis B vac-

    cination in hemodialysis patients. Among certain sub-groups of 

    older adults, influenza vaccination has also been linked to in-

    creased depressive symptoms, accompanied by a rise in serum lev-

    els of IL-6 (Glaser et al., 2003).

    8. Evidence for prediction # 3: elevated rates of infection and/or 

    immune alteration are found in depressed patients

     Just as elevated rates of infection would be expected among

    individuals with fever, elevated rates of infection and/or immunealteration are expected in depressed patients, given our proposal

    that depression is elicited as an immune defense during conditions

    of infection or immune-compromise.

    Accordingly, depressed individuals show marked evidence of 

    immune suppression and excessive inflammation (Dowlati et al.,

    2010; Irwin and Miller, 2007; Zorilla et al., 2001), as well as greater

    vulnerability to infection (Cohen, 1995; Irwin, 2002b; Zorilla et al.,

    1996). For example, a robust association has been found between

    depressive symptoms and reduced NK cell activity (Zorilla et al.,

    2001; Reiche et al., 2004). Depression has also been associated

    with decreased lymphocyte proliferation, although the results are

    less consistent, and appear to depend on moderating factors such

    as age (Schleifer et al., 1989).

    Increased levels of proinflammatory cytokines and chronic

    inflammatory response have been found repeatedly in depressed

    individuals (Dowlati et al., 2010; Irwin and Miller, 2007; Zorilla

    et al., 2001). Depressed individuals most often show evidence for

    increased levels of the proinflammatory cytokines IL-1b, IL-6,

    TNF-a, and INF-c (Maes et al., 2009). Other inflammatory biomark-ers that have shown regular associations with depressive symp-

    toms include changes in acute-phase-response protein, C-reactive

    protein (CRP), haptoglobin, nitric oxide, and glucocorticoids (Loftis

    et al., 2010; Maes et al., 2009; Zorilla et al., 2001 ).

    A number of studies indicate that depressed individuals show

    increased vulnerability to contracting infections, such as upper

    respiratory tract infections (Cohen, 1995). Among HIV-infected pa-

    tients, depressive symptoms are associated with higher viral load

    and lower NK cell activity (see review by  Kopinsky et al., 2004).

    Depressive symptoms are also significantly associated with recur-

    rence of herpes simplex infection (Zorilla et al., 1996) and reduced

    cellular immunity to varicella-zoster (Irwin, 2002b).

    9. Evidence for prediction # 4: many conditions associated with

    immune vulnerability are also associated with depression

    The list of conditions that show associations both with in-

    creased vulnerability to infection and with elevated risk for depres-sion is long and varied, and includes seasonal factors, nutritional

    deficiencies, hormonal fluctuations, toxin exposure, cancer, cardio-

    vascular disease, autoimmune diseases, and chronic pain (see Ta-

    naka et al., 2012, for a review). Two widely-studied factors linked

    both to immune vulnerability and to depression include stress

    and sleep deprivation. Stress, for example, which has a clear and

    well-established role in the etiology of depression (Brown et al.,

    1986; Hammen, 2005; Kendler et al., 1999; Sillaber et al., 2009),

    is also associated with increased numbers of circulating proinflam-

    matory cytokines (Bierhaus et al., 2003; Wolf et al., 2009). In-

    creased cytokines sensitize the HPA-axis, contributing to the

    hypersecretion of cortisol (Turnbull and Rivier, 1995), which has

    been identified as a critical pathway in central nervous system

    (CNS) dysregulation. Cortisol can suppress cellular immune re-sponse that is critical in defending the body against viral infections

    (Maes et al., 1994). Accordingly, there is evidence that chronic

    stress significantly increases vulnerability to infection (e.g., Cohen,

    1995; Cohen et al., 1999; Horesh et al., 2008; Kiecolt-Glaser et al.,

    2002; Mundt et al., 2000). For example, it has been found that

    stressed caretakers are more vulnerable to infectious diseases (Kie-

    colt-Glaser et al., 2002), and populations that experience higher

    levels of social stress tend to have higher rates of mortality and

    morbidity from infectious disease (Weiss and McMichael, 2004).

    Another major risk factor for depression is chronic insomnia

    (Lustberg and Reynolds, 2000). Chronic sleep deprivation is associ-

    ated with increased immune vulnerability (Irwin, 2002a), and the

    severity of insomnia is negatively correlated with NK cell activity

    in both depressed and non-depressed groups (Zorilla et al., 2001;Irwin, 2002a). Poor sleep quality and insomnia can precipitate

    S. Anders et al. / Brain, Behavior, and Immunity 31 (2013) 9–22   15

  • 8/20/2019 Depression as an Evolutionary Strategy for Defense Against Infection

    8/14

    inflammatory processes, raising plasma cortisol, C-reactive protein

    (CRP), and proinflammatory cytokines (Motivala et al., 2005). In

    experiments with rats, chronic restriction of sleep causes depres-

    sion-like changes in both the sensitivity of neurotransmitter recep-

    tors and neuroendocrine reactivity to stress (Novati et al., 2008).

    In addition to stress and sleep deprivation, the list of conditions

    that are associated both with immune vulnerability and with

    depression is staggering and diverse. They include seasonal factors

    such as reduced daylight hours and colder temperatures during the

    winter months (Lam et al., 2004; Magnusson, 2000), nutritional

    factors such as deficiencies of Vitamin D or Omega-3 fatty acids

    (Borges et al., 2011; Ganji et al., 2010; Hibbeln, 2009; McNamara

    et al., 2010; Parker et al., 2006), hormonal fluctuations associated

    with the menstrual cycle and post-partum period (Groër and Mor-

    gan, 2007; Rubinow et al., 2009), exposure to environmental toxins

    such as toxigenic mold and pesticides (Anyanwu et al., 2003; Bes-

    eler and Stallones, 2008; Corsini et al., 2008; Udoji et al., 2010), and

    a number of medical or physiological conditions such as cancer

    (Miller et al., 2008), cardiovascular disease (Cesari et al., 2003;

    Kling et al., 2007), autoimmune diseases (Gold and Irwin, 2006;

    Nery et al., 2008), and chronic pain (Watkins and Maier, 2000).

    That such a wide array of conditions has well-established links

    with both depression and immune alteration is consistent with

    the notion that depression evolved as a generalized immune re-

    sponse that can offer broad defense against a range of immune

    challenges. A preponderance of cross-sectional research in these

    areas limits causal interpretations and highlights the need for more

    longitudinal investigations; however, a number of experimental

    studies have reliably demonstrated that conditions associated with

    sources of immune vulnerability such as stress and sleep depriva-

    tion increase risk of depression-like behavior in animal models

    (e.g.,  Ardayfio and Kim, 2006; Novati et al., 2008; Sillaber et al.,

    2009).

    10. Evidence for prediction # 5: bidirectional links between the

    nervous and immune systems allow moods and immune vulnerability to influence each other 

    Multiple pathways have been identified by which immune sys-

    tem activation may lead to depression, including activation of the

    HPA-axis by proinflammatory cytokines (Holsboer, 2000; Pace

    et al., 2007) and degradation of tryptophan by activation of the

    IDO (Capuron et al., 2003; Dantzer et al., 2008). There is now

    well-established evidence to suggest that bidirectional communi-

    cation between the immune system and nervous system also oc-

    curs (Maier and Watkins, 1998).

    Bidirectional communication between the immune and nervous

    systems is suggested by the immune-enhancing effects of anti-

    depressant interventions, such as electroconvulsive therapy (ECT)

    (Fischler et al., 1992; Kronfol et al., 2002; Hestad et al., 2003), avariety of psychosocial interventions (Fang et al., 2010; Robinson

    et al., 2003; Witek-Janusek et al., 2008), and meditation practice

    (Lavretsky et al., 2012; Pace et al., 2009). For example, while

    ECT—known for its rapid amelioration of depressive symptoms—

    shows no evidence of changes in monoamine metabolism or neu-

    roplasticity following a single ECT session, augmented NK cell

    activity is found after a single session (Fischler et al., 1992; Kronfol

    et al., 2002), with a gradual and significant decline in TNF-a  levelsover the course of repeated sessions (Hestad et al., 2003).

    There is evidence that NK cell activity is significantly elevated

    after experimental exposure to pleasurable conditions (Berk

    et al., 2001; Matsunaga et al., 2008), and that psychological inter-

    ventions for the treatment of depression and emotional distress,

    such as mindfulness training and cognitive behavior therapy(CBT), also enhance NK cell activity (Masuda et al., 2002; Robinson

    et al., 2003; Witek-Janusek et al., 2008). Notably, among partici-

    pants in a mindfulness-based stress reduction program,   Fang

    et al. (2010) found that NK cell activity was significantly enhanced

    only in patients who reported improved well-being following the

    intervention.

    Studies of meditation practice also show notable effects on

    stress and immune responses. For example,   Pace et al. (2009)

    found that among individuals undergoing training for compassion

    meditation, the amount of time spent in meditation practice was

    associated with decreased IL-6 concentrations and decreased levels

    of distress following exposure to a laboratory stressor. In another

    study (Lavretsky et al., 2012), dementia caregivers who practiced

    daily yogic meditation experienced a reduction in depressive

    symptoms and reduced signs of stress-induced aging, measured

    by telomerase activity.

    Several types of antidepressants and mood stabilizers are also

    noted to have antibiotic effects. In a recent review,   Lieb (2007)

    cites a number of  in vivo   and in vitro  studies demonstrating that

    antidepressant drugs and lithium have significant antimicrobial

    and immune-potentiating effects. These effects include the ability

    to reverse resistance of bacteria to antibiotics (e.g.,   Kristiansen

    et al., 2010). It has been proposed that psychotropic medications

    derive their antimicrobial properties by acting as bacterial efflux

    pump inhibitors (EPIs) (Munoz-Bellido et al., 2000).   Lieb (2007)

    also notes that several antibiotics have been found to have

    mood-enhancing effects.

    11. Evidence for prediction # 6: moods orchestrate the timing 

    and intensity of infection-defense behaviors

    There is evidence that moods also provide a mechanism for reg-

    ulating the timing and intensity of immune-related behaviors

    based on an implicit cost-benefit analysis of what is most adaptive

    for survival and reproduction. Depressive behaviors are biologi-

    cally costly, interfering with work and social functioning, and espe-

    cially in pre-modern times would likely interfere with efforts toacquire and compete for food, territory, and mates. Therefore, the

    benefits would outweigh the costs only when initial efforts to cope

    with an immune challenge are ineffective, or when an individual

    suffers from chronic infection or stress. Sickness behavior and

    depression (and perhaps various sub-types of depression) are elic-

    ited—and vary in timing and intensity—based on feedback from the

    immune system. An extension of this is that mood elevation can

    stimulate more adventurous or gregarious behavior when an indi-

    vidual is in a more robust immune state.

    Evidence from a number of studies indicates that risk for

    depression and depression severity varies in relation to immune-

    compromise. For example, as mentioned above,   Murray et al.

    (2007)  found a heightened risk for depression among a group of 

    individuals who had contracted WNV disease, while those whohad been diagnosed with the more severe form of WNV, neuroin-

    vasive disease, demonstrated an even greater risk for depression.

    In a study of HIV patients, high viral loads were associated with

    higher depression scores on the Hamilton Rating Scale for Depres-

    sion (Cohen et al., 2002). Maes et al. (1994, 1995) have also found

    depression severity to correlate with circulating numbers of proin-

    flammatory cytokines, including IL-1 and IL-6, as well as with mea-

    sures of HPA-axis hyperactivity.

    There is also evidence that the timing of mood response to anti-

    depressant interventions parallels changes in immune function

    (Lieb, 2007; Tanaka and Kinney, 2011b). In a recent paper,  Tanaka

    and Kinney (2011b)   reviewed studies that provided information

    about the time course of mood response and NK cell activity fol-

    lowing a number of mood-enhancing interventions such as exer-cise, therapeutic administration of ketamine, and antidepressant

    16   S. Anders et al. / Brain, Behavior, and Immunity 31 (2013) 9–22

  • 8/20/2019 Depression as an Evolutionary Strategy for Defense Against Infection

    9/14

    treatments. Notable parallels in the time course of mood improve-

    ment and immune response were observed across all modalities.

    For example, both mood and NKCA are rapidly enhanced in re-

    sponse to exercise, and both of these effects tend to dissipate with-

    in several hours after the conclusion of exercise, following a similar

    time course. In a separate review,  Lieb (2007) described evidence

    that antidepressant drugs and lithium demonstrate concomitant

    increases in mood-response and immune-potentiating effects.

    12. Comparison of the infection-defense hypothesis with other 

    evolutionary theories of depression

    Although the proliferation of recent evidence linking depression

    to inflammation and immune function has led some to speculate

    about the potentially adaptive role of depression in helping fight

    infections, there is currently little in the way of systematic at-

    tempts to help explain depression from an evolutionary infec-

    tion-defense point of view. An exception to this is   Raison and

    Miller’s (2012)   recent hypothesis of pathogen host defense

    (PATHOS-D). Drawing on evidence that many genes identified as

    risk alleles for depression are also associated with immune factors

    related to pathogen host defense—such as hyperthermia, reduced

    bodily iron stores, conservation/withdrawal behavior, hypervigi-

    lance, and anorexia—PATHOS-D, like the infection-defense hypoth-

    esis, suggests that the advantages in pathogen host defense

    conferred by depression risk alleles offset depression’s notable

    costs to reproduction and survival. The two hypotheses differ,

    however, in their explanations of how depression relates to im-

    mune activation. According to PATHOS-D, genes that are involved

    in immune response and depression are one in the same, and

    depression is intrinsically tied to immune activation. The infec-

    tion-defense hypothesis, on the other hand, can be theoretically

    de-coupled from immune activation, suggesting that well-estab-

    lished mechanisms for communication between the immune sys-

    tem and the nervous system provide pathways for depression to

    be triggered and modulated by infections as well as other signalsof vulnerability, which is consistent with evidence that depression

    is commonly—but not always—associated with an immune-inflam-

    matory response (Raison and Miller, 2011).

    More traditional attempts to explain the persistence of depres-

    sion from an evolutionary standpoint have focused on socially

    adaptive mechanisms. Evolutionary theorists have often drawn

    upon key features of depression—such as sadness and with-

    drawal—as part of a subversive, ‘‘second-line’’ posture that offers

    protective advantages in response to defeat, and helps to explain

    many of depression’s puzzling, self-limiting features. For example,

    according to ‘rank theory,’ depression signals yielding behavior

    following an unsuccessful struggle for dominance in order to

    avoid unnecessary harm (Gilbert, 1992). Similarly, ‘social risk the-

    ory’ describes depression as a signal of submissiveness in re-sponse to social failure, as a way to maintain social ties critical

    for survival and reproduction (Allen and Badcock, 2006; Price

    et al., 1994). The ‘psychic pain hypothesis,’ offers a more general

    model of depressed mood, suggesting that it provides affective

    feedback that discourages continued investment in adverse or

    unreachable goals (Thornhill and Thornhill, 1989). Other theories

    describe ways in which behavioral changes and nonverbal cues

    associated with depression function as distress signals to elicit

    help during adversity (Hagen, 2003). Each theory hones in on

    selection pressures that have likely shaped affective responses

    throughout evolution; sadness, for example, heightening attention

    to adversity, provides important feedback to self and others about

    social and environmental sources of stress, and can promote adap-

    tive behavior in response to the types of losses or conflicts de-scribed above. From our infection-defense perspective, the

    immune system co-opts this affective response so that the deep,

    pervasive sense of sadness that often occurs in the context of clin-

    ical depression may help to promote withdrawal behaviors that

    conserve energy and reduce exposure to new sources of stress

    or infection.

    Traditional evolutionary theories of depression, while offering

    potentially useful explanations for understanding changes in af-

    fect and behavior under a circumscribed set of conditions, on

    the whole remain limited (1) in their ability to offer predictions

    that can be submitted to rigorous testing; (2) in their ability to ex-

    plain the full range of signs and symptoms of depression, includ-

    ing apparently paradoxical features such as hypervigilance vs.

    conservation/withdrawal states; (3) in their ability to explain

    depression’s association with a diverse array of conditions and ill-

    nesses such as nutritional deficiencies, seasonal changes, hor-

    monal fluctuations, and chronic diseases; and (4) in their failure

    to account for evidence that depression is linked to immune-

    inflammatory factors, mediated by a variety of neuroimmune pro-

    cesses. The infection-defense hypothesis, by contrast, helps to

    integrate a large and growing body of research that depression

    is intimately linked with immune function, and offers insight into

    the mechanisms by which many known risk factors for depres-

    sion, such as stress and sleep deprivation, contribute to the etiol-

    ogy of depression.

    13. Potential challenges to the infection-defense hypothesis

    Depression’s associations with higher morbidity and mortality

    rates for a number of diseases appear to contradict the notion that

    depression provides an evolutionary advantage. From the infec-

    tion-defense point of view, however, we would expect depression

    to be elicited more often in the context of illness and disease; thus,

    there would be extremely high rates of morbidity and mortality

    associated with it. There is some evidence to suggest that depres-

    sion not only follows, but also presages worse outcomes for a num-

    ber of illnesses, including HIV (Leserman, 2008) and cancer (Reicheet al. 2004). More longitudinal data is needed to provide an accu-

    rate account of the progression and relative contributions of 

    depression, inflammation, and potential ‘‘third variables’’ that

    may be influencing immune response. It is important to note, how-

    ever, that our hypothesis only requires that the relative advantages

    of depression outweigh its costs. Throughout human history, the

    impact of infection on survival and reproduction has been stagger-

    ing, and life expectancy throughout most of human history was

    less than half of what it is today; thus, immune responses are likely

    to be biased toward surviving acute or relatively short-term im-

    mune challenges, while the long-term effects of chronic immune-

    activation or inflammation only become more relevant as life

    expectancy gradually increases over time. Moreover, from a Dar-

    winian perspective, the fact that depression has persisted despitethese costs only strengthens the case that it must have offered a

    powerful adaptive advantage for risk alleles to have remained so

    high in the population.

    There may be other ways to understand depression’s persis-

    tence from an immune standpoint. For example, might depression

    simply be an epiphenomenon of a prolonged or severe inflamma-

    tory response? Or is it possible that depression is a maladaptive

    expression of genes that confer immune advantages to the host un-

    der some circumstances, and confer risk in others? Just as fever, a

    key asset in combatting infections, can be harmful or even lethal at

    high levels, an adaptive view of depression does not imply that

    depression can’t sometimes be harmful. However, the view that

    depression is a fundamentally maladaptive state is again refuted

    by the notably high prevalence rates of depression risk alleles inthe population.

    S. Anders et al. / Brain, Behavior, and Immunity 31 (2013) 9–22   17

  • 8/20/2019 Depression as an Evolutionary Strategy for Defense Against Infection

    10/14

    14. A comprehensive view of depression

    A comprehensive view of depression takes into account its mul-

    ti-faceted nature that manifests in a variety of clinical presenta-

    tions. For example, as noted above, some depressed individuals

    experience hypervigilant states that include agitation and restless-

    ness, while others experience low energy and psychomotor slow-

    ing (American Psychiatric Association, 2000). At any given pointin time, the specific features of depression that are evoked likely

    depend on the interaction of genetic factors (Raison and Miller,

    2012), early experience (Bradley et al., 2008; Chapman et al.,

    2004; Garnefski et al., 1990), immune vulnerability (Dantzer

    et al., 2008; Kinney and Tanaka, 2009), physiological vulnerabili-

    ties (Maes et al., 2009; Raison and Miller, 2011), and situational

    triggers (Keller and Nesse, 2006). For example, grief-related

    depression following the loss of a loved one is more likely to elicit

    crying, whereas depression that follows from stress or seasonal

    factors is more likely to involve pessimism and fatigue (Keller

    and Nesse, 2006). While not all individuals who experience depres-

    sion show evidence of increased inflammation (Raison and Miller,

    2011), it remains to be seen whether some forms of depression are

    better explained from alternate bio-behavioral frameworks. For

    example, grief reactions to separation and loss, rooted in survival

    strategies employed during the vulnerable state of infancy, may

    be better explained from an ethological attachment framework

    (Bowlby, 1982; Mikulincer and Shaver, 2007).

    Gene-environment interactions have been found to exert influ-

    ence on depression risk, as in the case of the 5-HTTLPR short allele

    of the serotonin transporter gene. Numerous studies have shown,

    for example, that carriers of the 5-HTTLPR short allele demonstrate

    an elevated risk for depression that is further increased in relation

    to levels of environmental stress (Eley et al., 2004; Kendler et al.,

    2005).

    Evidence from studies using both human and laboratory ani-

    mals suggest that early life experiences can influence susceptibility

    to inflammation and depression. Early adverse experiences such as

    neglect and trauma are related to increased risk for depression anddisease across the lifespan (Chapman et al., 2004; Garnefski et al.,

    1990). Childhood maltreatment is also associated with increased

    inflammation (Danese et al., 2007), and adults with a history of 

    childhood maltreatment have been found to respond to acute

    stress with increased IL-6 concentrations compared to controls

    (Carpenter et al., 2010). Laboratory studies using rats have found

    that impaired maternal contact and milk quality can induce signif-

    icant changes in stress response and inflammation in offspring

    (Walker, 2010). Increased CRH concentrations and HPA-axis activ-

    ity are found in both humans and laboratory animals who have

    experienced early life stress, and have been implicated as key fac-

    tors mediating the associations between early experience, inflam-

    mation, and depression (Bradley et al., 2008; Gillespie et al.,

    2009; Walker, 2010).Given the complex interplay of multiple systems involved in a

    depressive response, immune response may also interact with

    other physiological vulnerabilities to increase risk for depression.

    Maes and colleagues (Maes et al., 2009), for example, have outlined

    factors related to the condition of increased gut permeability,

    known as ‘‘leaky gut,’’ that may contribute to inflammation-associ-

    ated depression, such as increased translocation of lipopolysaccha-

    ride (LPS) from gram-bacteria.

    While the infection-defense hypothesis posits a protective role

    for depression in humans’ environment of evolutionary adapted-

    ness, the role of increased hygiene and availability of antimicrobial

    treatments in modern society have reduced mortality rates associ-

    ated with infection, and suggests the possibility that depression’s

    relative advantages in infection-defense may be decreasing. Thefact that rates of depression are increasing does not necessarily

    counter this, as the increasing rates may not be due to increased

    genetic risk per se, but rather to the increased presence of im-

    mune-compromising factors such as environmental toxins.

    It is also of note that, from a clinical perspective, psychological

    interpretations of depression—including psychodynamic or sche-

    ma-based interpretations—insofar as they help to identify and alle-

    viate sources of psychological stress, including classically-

    conditioned stress responses to situational triggers, complement

    an infection-defense view of depression. The infection-defense

    view of depression not only helps to provide an evolutionary

    explanation for the prevalence and persistence of depression

    throughout human history, but also supports an integrative frame-

    work for understanding the etiology of depression from multiple

    levels of influence.

    15. Summary and further clinical implications of the infection-

    defense hypothesis

    Converging evidence suggests that depression is often an

    inflammatory/immune-mediated response to infection, vulnerabil-

    ity to infection, and/or chronic activation of the innate immune

    system. This inflammatory response is stimulated by increasedproduction of proinflammatory cytokines, which have wide-rang-

    ing effects on both neuroendocrine and neuronal systems, includ-

    ing an inhibitory influence on serotonergic transmission. This

    emergent model of depression helps to explain: (1) why depres-

    sion is associated with immune alterations such as decreased

    NKCA, (2) why depression is associated with increased rates of 

    infection and disease, and (3) why a wide range of environmental

    and physiological factors associated with increased vulnerability to

    infection are also associated with increased risk for depression.

    In addition, the discovery of inflammatory-immune factors in

    the physiology of depression helps to explain an important psychi-

    atric puzzle as to why genes associated with major depression

    have persisted, despite depression’s association with increased

    morbidity and mortality. As outlined in the infection-defensehypothesis, signs and symptoms of depression such as anhedonia,

    social withdrawal, reduced energy and psychomotor retardation—

    and the genes that contribute to them—may be explained as adap-

    tive responses to infection vulnerability that serve to: (1) conserve

    metabolic resources for fighting infection, (2) reduce exposure to

    further infections or environmental stressors, and (3) reduce social

    contact to prevent the spread of infection to kin.

    Unlike many other evolutionary theories of depression, key te-

    nets derived from the infection-defense hypothesis can be submit-

    ted to rigorous testing. For example, using a double-blind placebo-

    controlled experimental design, a stringent test of the hypothesis

    that interventions reducing infection and/or immune compromise

    will lead to a reduction in depressive symptoms could be made. Fu-

    ture studies using longitudinal and epidemiological data will also

    be important in helping to establish the relative progression of im-

    mune-inflammatory factors and depression following exposure to

    infection or immune-compromising conditions.

    15.1. Clinical Implications

    Associations between immune function and depression, and in

    particular, the notion that moods may serve as a behavioral de-

    fense against infection, carry important implications for under-

    standing the causes, treatment, and prevention of depression.

    Some of these implications pose a challenge to conventional wis-

    dom; for example, if symptoms of depression are serving a protec-

    tive function in the face of immune challenge, then the goal of 

    reducing depressive symptoms to ease emotional distress shouldbe balanced against the need to first identify and treat possible

    18   S. Anders et al. / Brain, Behavior, and Immunity 31 (2013) 9–22

  • 8/20/2019 Depression as an Evolutionary Strategy for Defense Against Infection

    11/14

    underlying immune factors. Some factors that influence immune

    vulnerability, such as stress and sleep disturbances, are commonly

    addressed in mental health treatment settings and can be allevi-

    ated with traditional forms of therapy including CBT and mind–

    body relaxation techniques, as well as pharmacological interven-

    tions; however, the potential role of underlying infections and im-

    mune disorders warrants greater attention. Underlying infections

    or immune disorders may be under-diagnosed, and should rou-

    tinely be ruled out as potential causative factors. This need is

    underscored by the findings of  Rabinowitz et al. (1997)  that psy-

    chiatric patients tend to have higher rates of unrecognized and un-

    treated physical illness than the rest of the population.

    The notion that depressed individuals may have increased im-

    mune vulnerability, an underlying infection, or other source of im-

    mune compromise leads to several further implications. First,

    increased attention to hygiene in clinical settings such as waiting

    rooms, shared bedrooms on inpatient units, or other shared spaces

    may help to reduce the spread of pathogens that contribute to

    depression. Relatedly, depressed individuals may benefit from tak-

    ing heightened precautionary measures to reduce the risk of incur-

    ring further immune challenge, such as more thorough and

    frequent handwashing, or by avoiding crowded public places

    where there is greater exposure to pathogens. Finally, engaging

    in strategies to enhance immune function could potentially com-

    plement traditional anti-depressant therapies. The reduction of im-

    mune-compromising factors—such as sleep disorders, chronic pain,

    stress, dietary deficiencies, and insufficient exercise—deserves

    more investigation as a possible approach to treating and prevent-

    ing depression. For example, moderate exercise, meditation prac-

    tice, and nutritional supplementation to correct for deficiencies

    in omega-3 fatty acids and vitamin D, may be simple and econom-

    ical ways to help alleviate depressive symptoms.

    Environmental pathogens that trigger immune impairment and

    inflammation, such as toxigenic molds, pesticides, and other pollu-

    tants, also deserve greater attention for their potential role in the

    etiology of depression. For example, exposure to toxigenic mold

    is associated both with alterations in both NK cell activity and var-ious neuropsychiatric symptoms, including depression (Anyanwu

    et al., 2003). Increasing levels of environmental toxins that contrib-

    ute to inflammation and depression may potentially help to ex-

    plain recent increases in rates of depression (Compton et al., 2006).

    Pharmacologic interventions that have established antimicro-

    bial, immune-enhancing, or anti-inflammatory properties also

    merit more study for their potential antidepressant effects. For

    example, minocycline, a second-generation tetracycline antibiotic,

    has demonstrated anti-inflammatory and anti-depressant effects

    in both human and animal studies (Pae et al., 2008). Preliminary

    findings suggest that non-steroidal anti-inflammatory drugs (NSA-

    IDs) such as aspirin, ibuprofen, celecoxib, and naproxen may have

    antidepressant effects by way of their action as cyclooxygenase

    (COX)-2 inhibitors (Müller, 2010). For example, celecoxib has beenfound to block COX-2 enzymes as well as to reduce depression-like

    behaviors in laboratory animals (Guo et al., 2009). In at least one

    human study, celecoxib has been found to augment the anti-

    depressant effects of fluoxetine (Akhondzadeh et al., 2009). This

    latter finding, in particular, points to the possibility that COX-2

    inhibitors may be effective adjunct therapies to traditional

    antidepressants.

    Another promising line of research in the search for improved

    pharmacologic treatments of depression involves anti-inflamma-

    tory cytokine antagonists. For example, in animal studies, the use

    of the IL-1b receptor antagonist IL-1ra prevented the development

    of depression-like behavioral and neurochemical changes in re-

    sponse to chronic stress exposure (Koo and Duman, 2008; Norman

    et al., 2010). The anti-inflammatory cytokine IL-10 has also beenidentified as a potential target for antidepressant action. In studies

    with laboratory animals there is evidence that IL-10 modulates

    numerous features associated with depression including sleep,

    helplessness, and pain perception (Roque et al., 2009). Moreover,

    increased levels of circulating IL-10 have been found in both hu-

    mans and animals receiving anti-depressant treatment (Kenis

    and Maes, 2002).

    TNF-a   inhibitors have shown promise in clinical trials fordecreasing symptoms of depression. Patients with treatment-resis-

    tant depression who had high baseline levels of inflammatory bio-

    markers showed improvement following several infusions of the

    TNF antagonist Infliximab over a 12-week trial (Raison et al.,

    2012). Another TNF inhibitor, Etanercept, administered to psoriasis

    patients over a 12-week period, resulted in a greater decrease of 

    depressive symptoms from baseline when compared with placebo

    (Tyring et al., 2006).

    Of note, a potential risk of using anti-inflammatory agents in

    the treatment of depression is that the suppression of immune

    function may mask existing infections or increase susceptibility

    to new ones. This again underscores the need to rule out infection

    or immune-related causes of depression prior to treatment, and to

    weigh the potential risks of anti-inflammatory/immunosuppres-

    sant agents against the need to alleviate depressive symptoms.

    In summary, the infection-defense hypothesis bears important

    clinical implications for the treatment and prevention of depres-

    sion. Its basic premise that depression is an adaptive immune

    strategy (1) indicates that many behavioral features of depression

    may serve an adaptive purpose to help fight existing infections and

    avoid new ones; and (2) points to the need for a fundamental shift

    in depression treatment that favors the primacy of investigating

    and remedying underlying infections or immune-compromising

    factors (both environmental and endogenous) that may be contrib-

    uting to depression. In addition, interventions that aid the body’s

    natural ability to fight infections, such as correcting nutritional

    deficiencies or engaging in stress-reduction activities like medita-

    tion, would be expected, as numerous studies show, to help temper

    symptoms of depression. Finally, the proliferation of new discover-

    ies over the past several decades that link depression to immune-inflammatory processes, and that have identified bidirectional

    pathways by which the nervous and immune systems communi-

    cate, also opens the door for a wide range of novel pharmacologic

    interventions in the clinical management of depression.

    Conflict of interest

    All authors declare that there are no conflicts of interest.

    References

    Afsar, B., Elsurer, R., Eyileten, T., Yilmaz, M.I., Caglar, K., 2009. Antibody response

    following hepatitis B vaccination in dialysis patients: does depression and

    quality of life matter? Vaccine 27, 5865–5869.Akhondzadeh, S., Jafari, S., Raisi, F., Nasehi, A.A., Ghoreishi, A., Salehi, B., Mohebbi-

    Rasa, S., Raznahan, M., Kamalipour, A., 2009. Clinical trial of adjunctive

    celecoxib treatment in patients with major depression: a double blind and

    placebo controlled trial. Depress. Anxiety 26, 607–611.

    Allen, N.B., Badcock, P.B., 2006. Darwinian models of depression: a review of 

    evolutionary accounts of mood and mood disorders. Prog.

    Neuropsychopharmacol. Biol. Psychiatry 30, 815–826.

    American Psychiatric Association, 2000. Diagnostic and statistical manual of mental

    disorders text revision (4th ed.). American Psychiatric Association, Washington,

    DC.

    Andréasson, A., Arborelius, L., Erlanson-Albertsson, C., Lekander, M., 2007. A

    putative role for cytokines in the impaired appetite in depression. Brain

    Behav. Immun. 21, 147–152.

    Anyanwu, E., Campbell, A.W., Jones, J., Ehiri, J.E., Akpan, A.I., 2003. The neurological

    significance of abnormal natural killer cell activity in chronic toxigenic mold

    exposures. Sci. World J. 3, 1128–1137.

    Ardayfio, P., Kim, K., 2006. Anxiogenic-like effect of chronic corticosterone in the

    light-dark emergence task in mice. Behav. Neurosci. 120, 249–256.

    Avery, D.H., Shah, S.H., Eder, D.N., Wildschiødtz, G., 1999. Nocturnal sweating andtemperature in depression. Acta Psychiatr. Scand. 100, 295–301.

    S. Anders et al. / Brain, Behavior, and Immunity 31 (2013) 9–22   19

  • 8/20/2019 Depression as an Evolutionary Strategy for Defense Against Infection

    12/14

    Berk, L.S., Felten, D.L., Tan, S.A., Bittman, B.B., Westengard, J., 2001. Modulation of 

    neuroimmune parameters during the eustress of humorassociated mirthful

    laughter. Alter. Ther. Health Med. 7 (62–72), 74–76.

    Beseler, C.L., Stallones, L., 2008. A cohort study of pesticide poisoning and

    dep